Purpose: To develop and validate a population pharmacokinetic model for troxacitabine, a novel l-nucleoside analogue, administered by short infusion; to characterize clinical covariates that influence pharmacokinetic variability; and to design a dosage rate for continuous infusion administration to achieve low micromolar concentrations, which may be more efficacious than shorter infusions.

Experimental Design: Plasma samples from 111 cancer patients receiving troxacitabine (0.12-12.5 mg/m2) as a 30-minute infusion in phase I trials were used to develop the model with NONMEM. Clinical covariates evaluated included creatinine clearance, body surface area, age, and sex. From the model, a troxacitabine dosage rate of 2.0 to 3.0 mg/m2/d was expected to achieve a target concentration of 0.1 μmol/L; plasma samples were obtained during the infusion from eight patients receiving troxacitabine as a 3-day infusion.

Results: Troxacitabine pharmacokinetics were characterized by a three-compartment linear model. The mean value for systemic clearance [interindividual variability (CV%)] from the covariate-free model was 9.1 L/h (28%). Creatinine clearance and body surface area accounted for 36% of intersubject variation in clearance. Troxacitabine 2.0 mg/m2/d (n = 3) and 3.0 mg/m2/d (n = 5) for 3 days produced mean ± SD end of infusion concentrations of 0.12 ± 0.03 and 0.15 ± 0.03 μmol/L, respectively.

Conclusions: Renal function and body surface area were identified as sources of troxacitabine pharmacokinetic variability. The population pharmacokinetic model model–derived dosage rates for continuous infusion administration successfully achieved predetermined target plasma concentrations. The present model may be used to optimize treatment with troxacitabine by developing a dosing strategy based on both renal function and body size.

Troxacitabine, (−)-2′-deoxy-3′-oxacytidine (Fig. 1), is a novel unnatural l-nucleoside analogue with in vitro and in vivo antitumor activity against human leukemias and a variety of solid tumors, including hepatocellular, ovarian, prostate, and renal carcinomas (17). Like the related natural d-nucleoside analogues cytarabine and gemcitabine, troxacitabine requires phosphorylation to its active triphosphate form by intracellular kinases, including deoxycytidine kinase (1, 2, 8). Troxacitabine produces cytotoxic activity through incorporation of the triphosphate anabolite by DNA polymerases, complete DNA chain termination, and inhibition of DNA replication (9, 10). Unlike the natural d-nucleoside analogues, troxacitabine's l-configuration is resistant to catabolism by cytidine deaminase, which represents an inactivation step for other nucleoside analogues (2, 8, 9, 11). Troxacitabine is removed intracellularly by apurinic/apyrimidinic endonuclease (APE1), an enzyme responsible for DNA repair and regulation of gene expression (2, 8, 9, 11). Nucleoside transporters are involved in the cellular uptake of cytarabine and gemcitabine, whereas troxacitabine seems to accumulate intracellularly by passive diffusion (9).

Fig. 1.

Chemical structure of troxacitabine and other pyrimidine nucleoside analogues.

Fig. 1.

Chemical structure of troxacitabine and other pyrimidine nucleoside analogues.

Close modal

In phase I and II trials, troxacitabine given as an i.v. infusion for 30 minutes daily for 5 days showed significant antineoplastic activity in patients with relapsed myeloid leukemias, with the principal toxicities being grade 3 or 4 stomatitis, hand-foot syndrome, and skin rash (1214). Troxacitabine was also evaluated as a 30-minute infusion in patients with advanced solid malignancies on various administration schedules, including daily for 5 days every 3 to 4 weeks, weekly for 3 of 4 weeks, and once every 21 days, where the dose-limiting toxicity was neutropenia, and the most frequent nonhematologic toxicities were skin rash and hand-foot syndrome (1517).

In phase I studies, the pharmacokinetic behavior of troxacitabine was characterized by a mean plasma clearance of 10 L/h, with ∼3-fold variability between patients, and renal excretion resulting in the elimination of 61% to 77% of the given dose as unchanged drug (1517). Plasma exposure toxicity assessments in patients with refractory leukemia receiving 30-minute infusions showed that elevated troxacitabine plasma area under the concentration-time curve was related to the severity of mucositis and hand-foot syndrome (13).

Recent preclinical data suggest that troxacitabine given as a continuous infusion may be more efficacious than intermittent infusions. In a HT-29 human xenograft model, prolonged exposure to low micromolar concentrations (≥0.1 μmol/L) of troxacitabine for 3 to 5 days leads to superior growth inhibition compared with single high-dose bolus infusions (18). Based on this data, troxacitabine given by continuous infusion is being evaluated in patients with refractory acute myeloid leukemia (19). The starting dosage rate was 10.1 mg/m2/d, which was determined using the present population pharmacokinetic model to achieve a minimum target plasma concentration of 0.4 μmol/L. The initial infusion duration was 3 days with an increase in infusion to 5 to 6 days. In the first 13 patients treated, two complete responses were observed. With these promising results, accelerated development of continuous infusion troxacitabine is planned. A parallel phase I study of continuous infusion troxacitabine is being done in patients with advanced solid malignancies. In this study, the starting dosage rate was 2.0 to 3.0 mg/m2/d for 3 days, which was expected to reach a lower target plasma concentration of 0.1 μmol/L (20).

Population pharmacokinetic analysis is a useful tool for identification of sources of pharmacokinetic variability during anticancer drug development and can aid in the design of alternative dosing regimens to enhance their efficacy and safety (21). The objective of this study was to develop and validate a population pharmacokinetic model for troxacitabine to characterize clinical covariates that influence troxacitabine pharmacokinetics and to design a continuous dosing regimen to achieve a target plasma concentration of 0.1 μmol/L for phase I evaluation in patients with advanced solid malignancies.

Patient population and plasma troxacitabine levels

Troxacitabine plasma concentration versus time data from 111 patients with cancer from four separate phase I studies were used (12, 1517). Ninety-two (83%) had advanced solid tumors, and 19 (17%) had advanced leukemia. All concentration versus time data were obtained after a single i.v. dose of troxacitabine (0.12-12.5 mg/m2) given as a 30-minute infusion. All patients provided written informed consent, and the study protocols were approved by the investigational review boards from the respective institutions.

Four covariates were considered in the analysis, including continuous [age, body surface area (BSA), and creatinine clearance] and categorical (sex) variables. BSA was calculated using Mosteller's formula: BSA = [height (cm) × weight (kg) / 3,600]0.5 (22). Creatinine clearance was estimated using the Crockcroft and Gault method and expressed in units of mL/min; entrance criteria to the clinical protocols included an estimated creatinine clearance of ≥45 mL/min (23). Plasma sampling was extensive; a mean of 12.6 samples (range, 9-16 samples) per patient were obtained between 4 minutes to 168.5 hours after the start of infusion. Troxacitabine was quantitated in plasma using a method involving high-performance liquid chromatography with tandem mass spectrometry detection and calibration curves ranging from 0.6 to 99.9 ng/mL, as previously described (12, 15, 16), and all concentrations were measured in the same laboratory.

Pharmacokinetic model

The population pharmacokinetic model for troxacitabine was developed in two steps: (a) covariate-free (base) model development and (b) covariate model development. Pharmacokinetic models were fitted to the data using the computer software programs NONMEM version V, level 1.1 (GloboMax LLC, Hanover, MD) and PDx POP version 1.1 (Globomax LLC). Xpose 3.1/S-PLUS 6.0 (Insightful Corporation, Seattle, WA) was used for graphical diagnostics and covariate screen.

Step 1: covariate-free (base) model development. The troxacitabine population pharmacokinetic model was built using two drug input methods: (a) actual dose (mg/h; model A) and (b) dose normalized to BSA (mg/m2/h; model B). Model B was explored because the dose of troxacitabine given was normalized to a patient's individual BSA. This approach would provide an independent assessment of the contribution of BSA to interindividual variability on troxacitabine clearance when compared with the covariate-free model for model A. The best structural pharmacokinetic model for troxacitabine was defined by fitting the data to predefined pharmacokinetic models provided in the NONMEM library. Linear compartmental pharmacokinetic models were evaluated because linear pharmacokinetic characteristics were noted in previous phase I trials (12, 15, 16). The pharmacokinetic model incorporated unknown intersubject and intrasubject variability and defined the number and form of η, a measure of intersubject variability, and εij, a measure of intrasubject or residual variability. Intersubject variability for each pharmacokinetic variable was modeled with the exponential function. Residual error models of the additive, proportional, exponential, and combination methods were evaluated for the best structural pharmacokinetic model. The computational, intense, first-order conditional estimation method with or without interaction (FOCE + INTER) was used (24, 25).

For model discrimination, the NONMEM objective function (−2 times the log-likelihood function or −2 log-likelihood) was used to calculate the Aikake Information Criterion = (−2 log-likelihood) + 2p, for each nonhierarchical model, where 2p is twice the number of pharmacokinetic variables appearing in the model. Diagnostic plots for observed versus predicted plasma concentrations, residuals versus predicted concentrations, and weighed residuals versus predicted concentration were also considered during model discrimination.

Step 2: covariate model development. Covariate models were developed separately for models A and B as described above. Each candidate covariate identified from the screen was evaluated for inclusion to the selected structural pharmacokinetic model in a stepwise fashion by adding covariates in the order of highest to lowest level of significant correlation. General additive models and tree models were used in this analysis as implemented in the Xpose software (26).

Continuous variables were evaluated with the following models on pharmacokinetic variables as described in the manual for the software program PDx Pop: linear additive effect, normalized by median weight, centered on median weight, and power or allometric. The order of selecting pharmacokinetic variables for covariate model building was systemic clearance, volumes of distribution, and then intercompartmental clearance constants. Covariates were included into the model provided that the run resulted in successful minimization and covariate run status, a minimum reduction in objection function value of 3.875 (χ2 distribution for 1 degree of freedom with α = 0.05), and a reduction in the intersubject variance of the associated pharmacokinetic variable, and the %RSE (relative SE of estimation) associated with each variable estimate was <50%.

A stepwise backward deletion procedure was done for both full covariate models. Covariates remained in the final population pharmacokinetic model when the removal of the covariate resulted in a ≥10.828 increase in the objective function (χ2 distribution for 1 degree of freedom with α = 0.001). The final covariate models were run with the FOCE + INTER method.

Statistical assessment of final population pharmacokinetic models

To determine the influence of covariates on model prediction, a predictive performance evaluation was done for pharmacokinetic variables derived from both group I and II final population models as described previously (27, 28). Briefly, prediction errors (bias) were determined for pharmacokinetic variables estimated by the covariate-free (PE-N) and the final covariate (PE) models. For all models, percentage prediction error (PEj or PE-Nj) was calculated as (TVPKj − PKj) / TVPKj × 100, where TVPKj is the typical (population) value for the pharmacokinetic variable for patient j, and PKj is the true value for the pharmacokinetic variable for patient j. The individual calculated PEj and PE-Nj of the pharmacokinetic variable was compared to see if the final population pharmacokinetic model had improved on predicting the respective pharmacokinetic variable. An improvement was defined as having the absolute value of PEj to be less than the absolute value of PE-Nj.

Validation of final population pharmacokinetic models

Validation of the final population pharmacokinetic models was undertaken using internal cross-validation techniques as recommended by the guidance document from the U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research Center for Biologics Evaluation and Research, Guidance for industry: population pharmacokinetics, February 1999 (http://www.fda.gov/cder/guidance/1852fnl.pdf) and described previously by Vozeh et al. (29).

Internal cross-validation. Briefly, 20 different data sets were generated by randomly removing the observed plasma concentrations of five to six subjects in each data set from the entire data set of 111 subjects, where data from each subject was removed only once. Each of the 20 data sets were then fitted with the final population pharmacokinetic models to provide individual predicted plasma concentrations from which an individual's standardized mean prediction error was calculated as previously described (29). The mean and 95% confidence interval for standardized mean prediction error were assessed; the model was considered to provide unbiased estimates for predicting troxacitabine plasma concentrations if the 95% confidence interval included 0.

Application of population pharmacokinetic model for the design of a continuous infusion regimen. One objective of the present study was to develop a population pharmacokinetic model to design a continuous dosing regimen to sustain a troxacitabine plasma concentration of 0.1 μmol/L for 3 days. Based on the final population pharmacokinetic models, a continuous infusion of troxacitabine 2.0 to 3.0 mg/m2/d was estimated to achieve a minimum target plasma concentration of 0.1 μmol/L, which is being evaluated in patients with advanced solid tumors (20). In this study, plasma samples were obtained at 4 hours after the start of infusion then daily at 24-hour intervals, with a final sample immediately before termination of the infusion, to determine if the target concentration of 0.1 μmol/L was achieved. Plasma troxacitabine concentrations for this study were measured using the same assay and laboratory used for the phase I studies (12, 1517).

The population distributions of values for the patient covariates evaluated in the pharmacokinetic models are summarized in Table 1. All but three patients had adequate renal function defined as calculated creatinine clearance of >45 mL/min.

Table 1.

Patient covariates

CovariateMean ± SDQuartiles
Symbol
Min25%Median75%Max
BSA (m2)* 1.92 ± 0.25 1.34 1.78 1.95 2.08 2.66 BSA 
Sex (male/female) 89/22      SEX 
Age (y) 59.8 ± 12.4 29 50.9 60.0 69.1 83 AGE 
Calculated creatinine clearance (mL/min) 81.8 ± 30.5 34.3 60.9 75.4 95.6 191 CLCR 
CovariateMean ± SDQuartiles
Symbol
Min25%Median75%Max
BSA (m2)* 1.92 ± 0.25 1.34 1.78 1.95 2.08 2.66 BSA 
Sex (male/female) 89/22      SEX 
Age (y) 59.8 ± 12.4 29 50.9 60.0 69.1 83 AGE 
Calculated creatinine clearance (mL/min) 81.8 ± 30.5 34.3 60.9 75.4 95.6 191 CLCR 

Abbreviations: min, minimum value; max, maximum value.

*

BSA was calculated using the Mosteller's formula: (weight × height / 3,600)0.5.

Calculated creatinine clearance was calculated using the Crockcroft and Gault method.

Step 1: covariate-free (base) model development

The best structural (covariate-free) pharmacokinetic model that described troxacitabine concentrations was a three-compartment linear model (NONMEM ADVAN 11 with TRANS4 translator subroutines) with a combination residual error model. Results of the covariate-free models are summarized in Tables 2 and 3, where the mean (interindividual variability, CV%) values for systemic clearance were 9.09 L/h (28%) and 4.78 L/h/m2 (24%) for models A and B, respectively. Based on assessment of interindividual variability between the two models, normalization of drug dose to BSA reduced variability on troxacitabine clearance by 14%.

Table 2.

Typical population pharmacokinetic variables, expressed as estimate (%RSE), obtained from the base model and final model (model A) with drug input using the actual dose (mg/h), and using first-order conditional estimation with interaction computation method

Base model AFinal model A
Covariate None CL ∼ CLCR + BSA 
  V1 ∼ BSA 
  V2 ∼ BSA 
  Q3 ∼ BSA 
Typical estimates TVCL (L/h) = THETA(1)  
 TVCL (L/h) = 4.8 × CLCR0.431 + (BSA − 1.95) × 2.84  
 TVV1 (L) = THETA(2) TVV1 (L) = 14.2 × (BSA / 1.95)1.97 
 TVV2 (L) = THETA(3) TVV2 (L) = 14.6 × (BSA / 1.95)1.07 
 TVV3 (L) = THETA(4) TVV3 (L) = THETA(4) 
 TVQ2 (L/h) = THETA(5) TVQ2 (L/h) = THETA(5) 
 TVQ3 (L/h) = THETA(6) TVQ3 (L/h) = 5.17 × (BSA / 1.95)1.35 
OFV 8,210 8,066 
TVCL (L/h) 9.09 (3) 9.20* (10) 
TVV1 (L) 13.4 (8) 14.2 (7) 
TVV2 (L) 13.9 (6) 14.6 (6) 
TVV3 (L) 89.3 (15) 89.2 (13) 
TVQ2 (L/h) 15.3 (9) 15.5 (9.0) 
TVQ3 (L/h) 4.93 (5) 5.17 (5) 
IIV of CL (%) 28 (17) 18 (17) 
IIV of V1 (%) 63 (22) 58 (18) 
IIV of V2 (%) 15 (190) NE 
IIV of V3 (%) 45 (41) 46 (31) 
IIV of Q2 (%) 35 (68) 34 (72) 
IIV of Q3 (%) 34 (30) 27 (37) 
Base model AFinal model A
Covariate None CL ∼ CLCR + BSA 
  V1 ∼ BSA 
  V2 ∼ BSA 
  Q3 ∼ BSA 
Typical estimates TVCL (L/h) = THETA(1)  
 TVCL (L/h) = 4.8 × CLCR0.431 + (BSA − 1.95) × 2.84  
 TVV1 (L) = THETA(2) TVV1 (L) = 14.2 × (BSA / 1.95)1.97 
 TVV2 (L) = THETA(3) TVV2 (L) = 14.6 × (BSA / 1.95)1.07 
 TVV3 (L) = THETA(4) TVV3 (L) = THETA(4) 
 TVQ2 (L/h) = THETA(5) TVQ2 (L/h) = THETA(5) 
 TVQ3 (L/h) = THETA(6) TVQ3 (L/h) = 5.17 × (BSA / 1.95)1.35 
OFV 8,210 8,066 
TVCL (L/h) 9.09 (3) 9.20* (10) 
TVV1 (L) 13.4 (8) 14.2 (7) 
TVV2 (L) 13.9 (6) 14.6 (6) 
TVV3 (L) 89.3 (15) 89.2 (13) 
TVQ2 (L/h) 15.3 (9) 15.5 (9.0) 
TVQ3 (L/h) 4.93 (5) 5.17 (5) 
IIV of CL (%) 28 (17) 18 (17) 
IIV of V1 (%) 63 (22) 58 (18) 
IIV of V2 (%) 15 (190) NE 
IIV of V3 (%) 45 (41) 46 (31) 
IIV of Q2 (%) 35 (68) 34 (72) 
IIV of Q3 (%) 34 (30) 27 (37) 

Abbreviations: NE, negligible; IIV, interindividual variability (% CV); CLCR, creatinine clearance.

*

Calculated from the medium creatinine clearance of 4.52 L/h and medium BSA of 1.95 m2.

Calculated from the medium BSA of 1.95 m2.

Table 3.

Typical population pharmacokinetic variables, expressed as estimate (% RSE), obtained from the base model and final model (Model B) with drug input based on dose normalized to BSA (mg/h/m2), using first-order conditional estimation with interaction computation method

Base model BFinal model B
Covariate None CL ∼ CLCR 
Typical estimates TVCL (L/h/m2) = THETA(1) TVCL (L/h) = 2.72 × CLCR 0.373 
 TVV1 (L/m2) = THETA(2) TVV1 (L/m2) = THETA(2) 
 TVV2 (L/m2) = THETA(3) TVV2 (L/m2) = THETA(3) 
 TVV3 (L/m2) = THETA(4) TVV3 (L/m2) = THETA(4) 
 TVQ2 (L/h/m2) = THETA(5) TVQ2 (L/h/m2) = THETA(5) 
 TVQ3 (L/h/m2) = THETA(6) TVQ3 (L/h/m2) = THETA(6) 
OFV 8,104 8,072 
TVCL (L/h/m24.78 (3) 4.77* (9) 
TVV1 (L/m27.04 (7) 7.03 (7) 
TVV2 (L/m27.58 (4.4) 7.47 (6) 
TVV3 (L/m249.4 (15) 47.2 (12) 
TVQ2 (L/h/m28.35 (9) 8.37 (9) 
TVQ3 (L/h/m22.6 (4) 2.61(5) 
IIV of CL (%) 24 (16) 20 (16) 
IIV of V1 (%) 58 (19) 59 (19) 
IIV of V2 (%) NE NE 
IIV of V3 (%) 42 (47) 44 (31) 
IIV of Q2 (%) 31 (84) 31 (79) 
IIV of Q3 (%) 28 (38) 28 (36) 
Base model BFinal model B
Covariate None CL ∼ CLCR 
Typical estimates TVCL (L/h/m2) = THETA(1) TVCL (L/h) = 2.72 × CLCR 0.373 
 TVV1 (L/m2) = THETA(2) TVV1 (L/m2) = THETA(2) 
 TVV2 (L/m2) = THETA(3) TVV2 (L/m2) = THETA(3) 
 TVV3 (L/m2) = THETA(4) TVV3 (L/m2) = THETA(4) 
 TVQ2 (L/h/m2) = THETA(5) TVQ2 (L/h/m2) = THETA(5) 
 TVQ3 (L/h/m2) = THETA(6) TVQ3 (L/h/m2) = THETA(6) 
OFV 8,104 8,072 
TVCL (L/h/m24.78 (3) 4.77* (9) 
TVV1 (L/m27.04 (7) 7.03 (7) 
TVV2 (L/m27.58 (4.4) 7.47 (6) 
TVV3 (L/m249.4 (15) 47.2 (12) 
TVQ2 (L/h/m28.35 (9) 8.37 (9) 
TVQ3 (L/h/m22.6 (4) 2.61(5) 
IIV of CL (%) 24 (16) 20 (16) 
IIV of V1 (%) 58 (19) 59 (19) 
IIV of V2 (%) NE NE 
IIV of V3 (%) 42 (47) 44 (31) 
IIV of Q2 (%) 31 (84) 31 (79) 
IIV of Q3 (%) 28 (38) 28 (36) 

Abbreviations: NE, negligible; IIV, interindividual variability (% CV); CLCR, creatinine clearance.

*

Calculated from the medium creatinine clearance of 4.52 L/h.

Step 2: covariate model development

During the covariate screen for model A, where drug input was the actual dose in units of mg/h, creatinine clearance and BSA were identified as two significant covariates for troxacitabine clearance, and BSA was identified as a covariate for the variables V1, V2, and Q3. During the model building process, the addition of creatinine clearance as a covariate on troxacitabine clearance decreased interindividual variability (CV%) on this variable from 28% to 21% (Fig. 2A), representing ∼25% of intersubject variance. The further addition of BSA as a covariate on clearance decreased the CV% from 21% to 18%, indicating that BSA accounted for 14% of interindividual variability on troxacitabine clearance. These results are consistent with those obtained from the base models, where interindividual variability on clearance was decreased by 14% when drug in put was based on drug normalized to BSA (model B) compared with drug input based on the actual dose (model A). The objection function value continued to decrease with the successive addition of BSA on V1, V2, and Q3, but interindividual variability for clearance and other variables were not reduced (Fig. 3A). The stepwise backward deletion process resulted in the removal of no covariates from the covariate model.

Fig. 2.

Unexplained interindividual variability on troxacitabine clearance (% CV) during covariate model building for (A) model A (drug input was actual dose, mg/h) and (B) model B (drug input was dose normalized to BSA, mg/m2/h). Abbreviations: CL, clearance; CLCR, creatinine clearance.

Fig. 2.

Unexplained interindividual variability on troxacitabine clearance (% CV) during covariate model building for (A) model A (drug input was actual dose, mg/h) and (B) model B (drug input was dose normalized to BSA, mg/m2/h). Abbreviations: CL, clearance; CLCR, creatinine clearance.

Close modal
Fig. 3.

Observed versus population model-predicted plasma troxacitabine concentrations for the final covariate models: (A) model A (drug input was actual dose, mg/h) and (B) model B (drug input was dose normalized to BSA, mg/m2/h). The solid line is the fit from linear least-squares regression; the dotted line is the line of identity.

Fig. 3.

Observed versus population model-predicted plasma troxacitabine concentrations for the final covariate models: (A) model A (drug input was actual dose, mg/h) and (B) model B (drug input was dose normalized to BSA, mg/m2/h). The solid line is the fit from linear least-squares regression; the dotted line is the line of identity.

Close modal

Table 2 presents troxacitabine typical population pharmacokinetic variables for the base model and final model for model A using the more computationally intense FOCE + INTER estimation method. From base to final model, the value for objection function value decreased by 144 (from 8,210 to 8,066), and the addition of covariates (creatinine clearance and BSA) accounted for 36% of unexplained variation on clearance (CV% decreased from 28% to 18%). The goodness of fit between observed and final model-predicted troxacitabine concentrations are shown in Fig. 3A (R2 = 0.8918).

During the covariate screen for model B, where drug input was dose normalized to BSA in units of mg/m2/h, creatinine clearance was identified as a covariate for troxacitabine clearance. The addition of creatinine clearance on troxacitabine clearance decreased interindividual variability (CV%) for this variable from 24% to 20% (Fig. 2B). Subsequent models included the addition of AGE on V1, creatinine clearance on Q2, and BSA on Q3, with a significant reduction in the objective function, but provided no further explanation of interindividual variability on troxacitabine clearance (Fig. 2B). In addition, when AGE was added as a covariate on V1, the %RSE for V1 increased from 10% to 55% and increasingly worsened with addition of covariates on other variables. Likewise, with the addition of creatinine clearance on Q2, the %RSE for Q2 increased from 13% to 42%, and the %RSE for interindividual variability on this variable increased from 20% to 250%. Similarly, the addition of BSA to Q3 resulted in worsened %RSE (>100%) for this variable.

Table 3 presents troxacitabine typical population pharmacokinetic variables for the base model and final model for model B. From base to final model, the value for objection function value decreased by 32 (from 8,104 to 8,072), and the addition of creatinine clearance as a covariate accounted for 18% of unexplained variation on clearance (CV% decreased from 24% to 20%). The goodness of fit between observed and final model-predicted troxacitabine concentrations are shown in Fig. 3B (R2 = 0.8823).

Statistical assessment of final population pharmacokinetic models

Table 4 summarizes the prediction errors associated with pharmacokinetic variables estimated from the covariate-free (PE-N) and two final covariate (PE) population models, and the percentage of patients whose pharmacokinetic variable value predictions were improved by the covariate models. Addition of covariates reduced the prediction error and the SD for all pharmacokinetic variables and similar prediction errors were noted for both covariate models. Predictions were improved for clearance in >50% of patients for both models A and B.

Table 4.

Prediction errors associated with covariate-free (PE-N) and final population models A and B with covariates (PE)

Pharmacokinetic variablePE-N, mean ± SDPE, mean ± SDPatients, % improved
Model A    
    CL −3.44 ± 26 −1.99 ± 25 55.0 
    V1 −16.88 ± 65 −9.90 ± 61 53.2 
    V2 0.16 ± 6.1 2.82 ± 12 26.1 
    V3−4.48 ± 34 −5.10 ± 35 40.5* 
    Q2−0.71 ± 19 −0.83 ± 19 58.6* 
    Q3 −4.18 ± 33 0.26 ± 29 45.0 
Model B    
    CL −3.28 ± 22 −2.49 ± 22 53.2 
    V1−15.1 ± 61 −15.0 ± 61 39.6* 
    V2   
    V3−4.24 ± 31 −3.30 ± 28 30.6* 
    Q2−0.25 ± 16 −0.17 ± 16 34.2* 
    Q3−2.65 ± 24 −2.50 ± 24 38.7* 
Pharmacokinetic variablePE-N, mean ± SDPE, mean ± SDPatients, % improved
Model A    
    CL −3.44 ± 26 −1.99 ± 25 55.0 
    V1 −16.88 ± 65 −9.90 ± 61 53.2 
    V2 0.16 ± 6.1 2.82 ± 12 26.1 
    V3−4.48 ± 34 −5.10 ± 35 40.5* 
    Q2−0.71 ± 19 −0.83 ± 19 58.6* 
    Q3 −4.18 ± 33 0.26 ± 29 45.0 
Model B    
    CL −3.28 ± 22 −2.49 ± 22 53.2 
    V1−15.1 ± 61 −15.0 ± 61 39.6* 
    V2   
    V3−4.24 ± 31 −3.30 ± 28 30.6* 
    Q2−0.25 ± 16 −0.17 ± 16 34.2* 
    Q3−2.65 ± 24 −2.50 ± 24 38.7* 

Abbreviations: CL, clearance; V1, volume of distribution of central compartment; V2, volume of distribution of peripheral compartment 1; V3, volume of distribution of peripheral compartment 2; Q2, intercompartment clearance constant between central and peripheral compartment 1; Q3, intercompartment clearance constant between central and peripheral compartment 2.

*

Pharmacokinetic variable with no covariate added.

Validation of final population pharmacokinetic models

Internal cross-validation. The mean (95% confidence interval) standardized mean prediction error for the final covariate model B was 0.0508 (−0.0553 to 0.157), suggesting that this covariate model provides unbiased estimates for predicting troxacitabine plasma concentrations (95% confidence interval contains 0). The correlation (R2) between observed and final model-predicted concentrations was 0.8775. Convergence was not achieved during internal cross validation of final covariate model A. Model A incorporated more variables than model B (13 versus 9), and overparameterization may have precluded convergence of model A.

Application of population pharmacokinetic model for the design of a continuous infusion regimen. Eight patients received troxacitabine 2.0 mg/m2/d (n = 3) for 3 days or 3.0 mg/m2/d for 3 days (n = 5). At 2.0 mg/m2/d, observed mean ± SD concentrations at 4, 24, 48, and 72 hours (end of infusion) were 0.06 ± 0.002, 0.10 ± 0.006, 0.10 ± 0.01, and 0.12 ± 0.03 μmol/L, respectively; at 3.0 mg/m2/d, concentrations were 0.08 ± 0.01, 0.13 ± 0.02, 0.15 ± 0.04, and 0.15 ± 0.03 μmol/L, respectively. Assessment of end of infusion concentrations show that the minimum target concentration of 0.1 μmol/L was achieved in all patients with the starting dose of 2.0 to 3.0 mg/m2/d, which was derived from the described population pharmacokinetic model.

This report provides the first description of a population pharmacokinetic model for troxacitabine, a novel unnatural l-nucleoside analogue, and the influence of clinical covariates on drug disposition. Renal function, assessed by calculated creatinine clearance, had the most significant effect on troxacitabine clearance. BSA, which is currently used to normalize troxacitabine dose, was also found to be an independent covariate on drug clearance. Creatinine clearance and BSA combined accounted for 36% of intersubject variance in troxacitabine clearance. Because elevated troxacitabine plasma exposure has been related to the severity of toxicities in phase I and II trials, including neutropenia, mucositis, and hand-foot syndrome, the described model has potential for optimization of troxacitabine therapy.

Troxacitabine is eliminated principally via renal excretion. During model development, it was anticipated that creatinine clearance would be a covariate explaining a significant percentage of interindividual variability in drug clearance. The addition of BSA as an independent, significant covariate on clearance is generally consistent with the correlation of body size with glomerular filtration rate (30) and supports the calculation of troxacitabine dose based on BSA. During covariate screening, age was not selected as a significant covariate on clearance. Because age was included in the formula for calculated creatinine clearance, inclusion of this variable may have accounted for age-related decline in renal function. Perhaps the inclusion of younger pediatric and older geriatric patients into the population may have resulted in retaining age into the final model. Sex was not included into the model most likely due to the predominance of male subjects (80%).

Both final models, one where drug input was the actual dose (model A) and one where drug input was the dose normalized to BSA (model B), equally accounted for a significant percentage of interindividual variability for troxacitabine clearance. Final estimates for interindividual variability were 18% and 20% for models A and B, respectively. SEs (%RSE) on variable estimates and predictive performance were similar for models A and B.

Recent preclinical data suggest that prolonged troxacitabine exposure at low micromolar concentrations (≥0.1 μmol/L) was superior to bolus infusion administration, and that a 3- to 5-day infusion achieved optimal efficacy (18). Troxacitabine penetrates into cells via passive diffusion, which may explain, in part, why troxacitabine showed enhanced cytotoxicity with prolonged versus bolus administration. To optimize the therapeutic potential of troxacitabine, several phase I trials were designed to evaluate prolonged infusion schedules (19, 20). The population pharmacokinetic model was used to derive a starting dosage rate to achieve a target troxacitabine plasma concentration of 0.1 μmol/L in patients with advanced solid malignancies, and the target plasma concentration was achieved in all patients receiving 2.0 to 3.0 mg/m2/d, albeit with accompanying intersubject pharmacokinetic variability. The equations for troxacitabine clearance as described in Tables 2 and 3 (final models), could be used to individualize the dosage rate for continuous infusion administration. For example, an individualized dose could be determined using values for creatinine clearance and BSA and the clearance equation in Table 2 according to the following equation: dose (μg/h) = [(4.8 × creatinine clearance0.431) + (BSA − 1.95) × 2.84] × target Css (μg/L). If the target Css is 0.1 μmol/L (21 μg/L), the individualized dosage rate for an individual with a BSA of 1.80 m2 and creatinine clearance of 45 mL/min (2.7 L/h) would be 146 μg/h (3.5 mg/d); the individualized dosage rate for an individual with the same BSA but a creatinine clearance of 100 mL/min (6.0 L/h) would be 209 μg/h (5.0 mg/d). If both patients received the same dose normalized to their BSA (e.g., 2.0 mg/m2/d or 3.6 mg/d), the patient with the higher creatinine clearance may have been undertreated. The clinical applicability and safety of the proposed dosing strategy would require prospective evaluation compared with the currently used BSA-based dosing.

In conclusion, a population pharmacokinetic model has been developed that accounted for 36% of unexplained intersubject variance for troxacitabine clearance using measures of renal function (calculated creatinine clearance) and body size (BSA). Further refinement of dosing strategies to decrease pharmacokinetic variability will likely include more accurate assessment of glomerular filtration rate and renal function. In addition, the population pharmacokinetic model was used to determine a dosage rate for continuous infusion administration that achieved predetermined target plasma concentrations. The present analysis shows that population pharmacokinetic modeling can be incorporated in the drug development process to identify sources of pharmacokinetic variability, aid in the design of alternative dosing regimens, and optimize therapy.

Grant support: Shire Pharmaceuticals.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Conflict of interest statement: S.D. Baker and M. Hidalgo received research funding from Shire Pharmaceuticals.

1
Grove KL, Guo X, Liu SH, Gao Z, Chu CK, Cheng YC. Anticancer activity of β-l-dioxolane-cytidine, a novel nucleoside analogue with the unnatural l configuration.
Cancer Res
1995
;
55
:
3008
–11.
2
Grove KL, Cheng YC. Uptake and metabolism of the new anticancer compound β-l-(−)-dioxolane-cytidine in human prostate carcinoma DU-145 cells.
Cancer Res
1996
;
56
:
4187
–91.
3
Kadhim SA, Bowlin TL, Waud WR, et al. Potent antitumor activity of a novel nucleoside analogue, BCH-4556 (β-l-dioxolane-cytidine), in human renal cell carcinoma xenograft tumor models.
Cancer Res
1997
;
57
:
4803
–10.
4
Siu LL, Attardo G, Izbicka E, et al. Activity of (−)-2′-deoxy-3′-oxacytidine (BCH-4556) against human tumor colony-forming units.
Ann Oncol
1998
;
9
:
885
–91.
5
Rabbani SA, Harakidas P, Bowlin T, Attardo G. Effect of nucleoside analogue BCH-4556 on prostate cancer growth and metastases in vitro and in vivo.
Cancer Res
1998
;
58
:
3461
–5.
6
Weitman S, Marty J, Jolivet J, Locas C, Von Hoff DD. The new dioxolane, (−)-2′-deoxy-3′-oxacytidine (BCH-4556, troxacitabine), has activity against pancreatic human tumor xenografts.
Clin Cancer Res
2000
;
6
:
1574
–8.
7
Gourdeau H, Bibeau L, Ouellet F, Custeau D, Bernier L, Bowlin T. Comparative study of a novel nucleoside analogue (Troxatyl, troxacitabine, BCH-4556) and AraC against leukemic human tumor xenografts expressing high or low cytidine deaminase activity.
Cancer Chemother Pharmacol
2001
;
47
:
236
–40.
8
Galmarini CM, Mackey JR, Dumontet C. Nucleoside analogues: mechanisms of drug resistance and reversal strategies.
Leukemia
2001
;
15
:
875
–90.
9
Gourdeau H, Clarke ML, Ouellet F, et al. Mechanisms of uptake and resistance to troxacitabine, a novel deoxycytidine nucleoside analogue, in human leukemic and solid tumor cell lines.
Cancer Res
2001
;
61
:
7217
–24.
10
Gunzburg WH, Salmons B. Novel clinical strategies for the treatment of pancreatic carcinoma.
Trends Mol Med
2001
;
7
:
30
–7.
11
Chou KM, Kukhanova M, Cheng YC. A novel action of human apurinic/apyrimidinic endonuclease: excision of l-configuration deoxyribonucleoside analogs from the 3′ termini of DNA.
J Biol Chem
2000
;
275
:
31009
–15.
12
Giles FJ, Cortes JE, Baker SD, et al. Troxacitabine, a novel dioxolane nucleoside analog, has activity in patients with advanced leukemia.
J Clin Oncol
2001
;
19
:
762
–71.
13
Giles FJ, Garcia-Manero G, Cortes JE, et al. Phase II study of troxacitabine, a novel dioxolane nucleoside analog, in patients with refractory leukemia.
J Clin Oncol
2002
;
20
:
656
–64.
14
Giles FJ, Feldman EJ, Roboz GJ, et al. Phase II study of troxacitabine, a novel dioxolane nucleoside analog, in patients with untreated or imatinib mesylate-resistant chronic myelogenous leukemia in blastic phase.
Leuk Res
2003
;
27
:
1091
–6.
15
de Bono JS, Stephenson J, Jr., Baker SD, et al. Troxacitabine, an l-stereoisomeric nucleoside analog, on a five-times-daily schedule: a phase I and pharmacokinetic study in patients with advanced solid malignancies.
J Clin Oncol
2002
;
20
:
96
–109.
16
Belanger K, Moore M, Baker SD, et al. Phase I and pharmacokinetic study of novel l-nucleoside analog troxacitabine given as a 30-minute infusion every 21 days.
J Clin Oncol
2002
;
20
:
2567
–74.
17
Canova A, Yee L, Baker SD, Jolivet J. A phase I and pharmacokinetic (PK) study of β-l dioxalo-cytidine (BCH-4556) administered weekly for three weeks every 28 days [abstract 759].
Proc Amer Soc Clin Oncol
1999
;
18
:
197a
.
18
Leblond L, Jolivet J, Dong K, et al. Antitumor efficacy of troxacitabine given by continuous administration: the human HT-29 colon xenograft used as a tumor model.
Proc Am Assoc Cancer Res
2003
;
44
:
518
.
19
Feldman EJ, Roboz GJ, Allen-Bard SL, et al. Phase I evaluation of troxacitabine (TroxatylTM) administered by continuous infusion in patients with refractory acute myeloid leukemia (AML).
Proc Am Soc Hematol
2003
;
102
:
873a
.
20
Hidalgo M, Lee CKK, Carducci K, Jolivet J, Baker SD. Phase I evaluation of troxacitabine administered by continuous infusion in patients with refractory solid tumors.
Proc Amer Soc Clin Oncol
2004
;
23
:
144a
.
21
Kastrissios H, Ratain MJ. Screening for sources of interindividual pharmacokinetic variability in anticancer drug therapy: utility of population analysis.
Cancer Invest
2001
;
19
:
57
–64.
22
Mosteller RD. Simplified calculation of body-surface area.
N Engl J Med
1987
;
317
:
1098
.
23
Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine.
Nephron
1976
;
16
:
31
–41.
24
Jonsson EN, Wade JR, Karlsson MO. Nonlinearity detection: advantages of nonlinear mixed-effects modeling.
AAPS PharmSci
2000
;
2
:
E32
.
25
Wahlby U, Jonsson EN, Karlsson MO. Assessment of actual significance levels for covariate effects in NONMEM.
J Pharmacokinet Pharmacodyn
2001
;
28
:
231
–52.
26
Jonsson EN, Karlsson MO. Xpose-an S-PLUS based population pharmacokinetic/pharmacodynamic model building aid for NONMEM.
Comput Methods Programs Biomed
1999
;
58
:
51
–64.
27
Bruno R, Vivler N, Vergniol JC, De Phillips SL, Montay G, Sheiner LB. A population pharmacokinetic model for docetaxel (Taxotere): model building and validation.
J Pharmacokinet Biopharm
1996
;
24
:
153
–72.
28
Gallo JM, Laub PB, Rowinsky EK, Grochow LB, Baker SD. Population pharmacokinetic model for topotecan derived from phase I clinical trials.
J Clin Oncol
2000
;
18
:
2459
–67.
29
Vozeh S, Maitre PO, Stanski DR. Evaluation of population (NONMEM) pharmacokinetic parameter estimates.
J Pharmacokinet Biopharm
1990
;
18
:
161
–73.
30
Peters AM, Henderson BL, Lui D. Indexed glomerular filtration rate as a function of age and body size.
Clin Sci (Lond)
2000
;
98
:
439
–44.